Synthesis of the Novel PARP-1 Inhibitor AG-690/11026014 and Its

Total Page:16

File Type:pdf, Size:1020Kb

Synthesis of the Novel PARP-1 Inhibitor AG-690/11026014 and Its Acta Pharmacologica Sinica (2017) 38: 638–650 © 2017 CPS and SIMM All rights reserved 1671-4083/17 www.nature.com/aps Original Article Synthesis of the novel PARP-1 inhibitor AG- 690/11026014 and its protective effects on angiotensin II-induced mouse cardiac remodeling Guo-shuai FENG1, Cui-ge ZHU2, Zhuo-ming LI1, Pan-xia WANG1, Yi HUANG1, Min LIU3, Ping HE1, Lan-lan LOU2, Shao-rui CHEN1, *, Pei-qing LIU1, * 1Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; 2Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; 3Department of Pharmacology, Shandong University School of Medicine, Ji-nan 250012, China Abstract We previously identified AG-690/11026014 (6014) as a novel poly(ADP-ribose) polymerase-1 (PARP-1) inhibitor that effectively prevented angiotensin II (Ang II)-induced cardiomyocyte hypertrophy. In the present study, we reported a new synthesis route for 6014, and investigated its protective effects on Ang II-induced cardiac remodeling and cardiac dysfunction and the underlying mechanisms in mice. We designed a new synthesis route to obtain a sufficient quantity of 6014 for this in vivo study. C57BL/6J mice were infused with Ang II and treated with 6014 (10, 30, 90 mg·kg-1·d-1, ig) for 4 weeks. Then two-dimensional echocardiography was performed to assess the cardiac function and structure. Histological changes of the hearts were examined with HE staining and Masson’s trichrome staining. The protein expression was evaluated by Western blot, immunohistochemistry and immunofluorescence assays. The activities of sirtuin-1 (SIRT-1) and the content of NAD+ were detected with the corresponding test kits. Treatment with 6014 dose- dependently improved cardiac function, including LVEF, CO and SV and reversed the changes of cardiac structure in Ang II-infused mice: it significantly ameliorated Ang II-induced cardiac hypertrophy evidenced by attenuating the enlargement of cardiomyocytes, decreased HW/BW and LVW/BW, and decreased expression of hypertrophic markers ANF, BNP and β-MHC; it also prevented Ang II-induced cardiac fibrosis, as implied by the decrease in excess accumulation of extracellular matrix (ECM) components collagen I, collagen III and FN. Further studies revealed that treatment with 6014 did not affect the expression levels of PARP-1, but dose-dependently inhibited the activity of PARP-1 and subsequently restored the activity of SIRT-1 in heart tissues due to the decreased consumption of NAD+ and attenuated Poly-ADP-ribosylation (PARylation) of SIRT-1. In conclusion, the novel PARP-1 inhibitor 6014 effectively protects mice against AngII-induced cardiac remodeling and improves cardiac function. Thus, 6014 might be a potential therapeutic agent for heart diseases.. Keywords: cardiac remodeling; PARP-1 inhibitor; 6014; PARylation; NAD+; SIRT-1 Acta Pharmacologica Sinica (2017) 38: 638–650; doi: 10.1038/aps.2016.159; published online 27 Feb 2017 Introduction an important component of the RAAS, could induce cardiac [2–6] Cardiac remodeling, manifested clinically as hypertrophy remodeling following sustained hypertension . (enlargement), fibrosis, and dysfunction of the heart resulting PARylation, induced by poly(ADP-ribose) polymerases from cardiac load or injury, is generally accepted as a deter- (PARPs), is a type of reversible posttranslational modifica - minant of the pathogenesis of heart failure (HF)[1]. The renin- tion impacting numerous cellular processes, eg, DNA repair, [7] angiotensin-aldosterone system (RAAS) plays a pivotal role in transcription, metabolism, or immune functions . PARP-1, the physiological and pathological processes of cardiac remod- the predominant member of the PARPs, accounts for approxi- [8] eling. It is also widely accepted that angiotensin II (Ang II), mately 90% of the cellular PARylation events . Once acti- vated, PARP-1 catalyzes protein PARylation, which results in the formation of polymerized ADP-ribose (PAR) dependently *To whom correspondence should be addressed. on NAD+ donor molecules, and subsequently the covalent E-mail [email protected] (Shao-rui CHEN); [9] [email protected] (Pei-qing LIU) attachment of PAR polymers to target proteins . Received 2016-09-26 Accepted 2016-11-24 PARP-1 is involved in pathological cardiac remodeling www.chinaphar.com Feng GS et al 639 and heart failure. Inhibition of PARP-1 prevents pathologi- eluted with a gradient of 5% to 95% solvent A for 25 min and cal cardiac remodeling induced by partial abdominal aortic detected at 254 nm; solvent A was CH3CN in double-deionized [10] 1 13 constriction . PARP-1-deficient mice are protected from Ang H2O. H NMR and C NMR spectra (AvanceIII, Burker) were II-induced cardiac hypertrophy[11]. Moreover, PARP-1 activa- recorded using TMS as an internal standard with a Burker - tion facilitates the pathogenesis of cardiac fibrosis[12] and the BioSpinUltrashield 400 NMR system. High-resolution mass development of heart failure[13, 14]. These findings indicate that spectra (HRMS) (LCMS-IT-TOF, Shimadzu) were recorded on PARP-1 could serve as a promising target for cardiac diseases, a Shimadzu LCMS-IT-TOF Instrument. and the pharmacologic inhibition of PARP-1 may be a poten- tial therapeutic strategy[15–17]. General procedure for the synthesis of 6014 A previous study from our laboratory identified AG-690/ (1) 4-chlorophenol (77.8 mmol), 2-(chloromethyl)oxirane (306.9 11026014(6014) as a novel PARP-1 inhibitor from the Specs mmol) and K2CO3 (231.5 mmol) were refluxed in acetone database (www.specs.net), which contains approximately (100 mL) overnight, cooled to room temperature and filtered. 200,000 compounds based on virtual screening. We also The filtrate was concentrated and extracted with toluene and reported that 6014 could significantly attenuate Ang II- water, and then the 2-((4-chlorophenoxy)methyl)oxirane (M1) induced cardiomyocyte hypertrophy[18]. To further deter - in toluene was purified by column chromatography. mine the in vivo cardiac protective effect of 6014, the present (2) 3-methyl-1H-purine-2,6(3H,7H)-dione (30 mmol) and study used C57BL/6J mice infused with Ang II as the model, AcONa (60 mmol) were dissolved in AcOH, and Br 2 was and elucidated the effects and underlying mechanisms of added dropwise and reacted for 2 h at 65 °C. 8-Bromo-3- this novel PARP-1 inhibitor. This study aimed to determine methyl-1H-purine-2,6(3H,7H)-dione (M2) was generated by whether 6014, as a PARP-1 inhibitor, has therapeutic potential cooling, filtering, water washing and drying in a vacuum. for cardiac remodeling. (3) M1 (31.0 mmol) and M2 (25.8 mmol) in ethanol (120 mL) were combined with trimethylamine (2.6 mmol) and refluxed Materials and methods for 3 h. 8-Bromo-7-(3-(4-chlorophenoxy)-2-hydroxypropyl)- Reagents 3-methyl-1H-purine-2,6(3H,7H)-dione (M3) was produced by The compounds 3-aminobenzamide (3-AB), 4’,6-diamidino- cooling, filtering, ethanol washing and drying in a vacuum. 2-phenylindole (DAPI) and Ang II were purchased from (4) M3 (8.15 mmol) and 2-mercaptoacetic acid (16.3 mmol) in Sigma-Aldrich (St Louis, MO, USA). Anti-PARP-1, anti- DMF (35 mL) were combined with NaHCO3 (32.6 mmol) and collagen I and anti-collagen III antibodies were provided reacted for 2 h at 85 °C. The reaction was stopped with water by Proteintech Group (Chicago, IL, USA). Anti-ANF and and then washed by methyl tertiary butyl ether. The products anti-FN antibodies were purchased from Santa Cruz Bio - in the water were combined with HCl (2.0 mmol) until the pH technology (Santa Cruz, CA, USA). Anti-BNP antibody was reached 3, and 2-((7-(3-(4-chlorophenoxy)-2-hydroxypropyl)-3- obtained from Merck Millipore (Bedford, Ohio, USA). Anti- methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio)acetic PAR antibody was obtained by Trevigen (Gaithersburg, MD, acid (M4) was yielded after filtering, water washing and dry- USA). Anti-SIRT-1, anti-lamin B1, anti-GAPDH antibodies ing in a vacuum. and all secondary antibodies for Western blot experiments (5) M4 (7.26 mmol) and ammonium chloride (21.7 mmol) were purchased from Cell Signaling Technology (Boston, in DMF (40 mL) was added to HATU (8.71 mmol) and DIEA MA, USA). An hematoxylin and eosin (HE) staining kit was (21.8 mmol) on ice and reacted overnight at room tempera- purchased from Santa Cruz Biotechnology (Santa Cruz, CA, ture. The products were precipitated by adding water and USA). A Masson’s staining kit and immunohistochemistry then filtering. 2-((7-(3-(4-chlorophenoxy)-2-hydroxypropyl)- reagents including Cy3-conjugated goat anti-mouse IgG (H+L) 3-methyl-2,6-dioxo-2,3,6,7-tetrahydro-1H-purin-8-yl)thio) were purchased from Wuhan Goodbio Technology Co, Ltd acetamide (6014) was purified by preparative chromatography (Wuhan, China). RIPA lysis buffer and IP lysis buffer were (Varian, Agilent). purchased from Beyotime Biotechnology (Jiangsu, China). The The synthesized intermediates M1-M4 were structurally protease and phosphatase inhibitor mini tablets, BCA protein identified by 1H NMR or MS, and 6014 was structurally identi- assay kit, Bradford protein assay kit, and protein G-agarose fied by 1H NMR, 13C NMR and HRMS. beads were obtained from Pierce Biotechnology (Rockford, AL, USA). A nuclear extract kit was purchased from ACTIVE 2-((4-chlorophenoxy)methyl)oxirane (M1) 1 MOTIF (Carlsbad, NM, USA). The quick start Bradford dye Crystal oil, yield 62.0%; H NMR (400 MHz, CDCl3) δ 7.28–7.22 reagent was obtained from Bio-Rad (Richmond, VA, USA). A (m, 2H), 6.90–6.83 (m, 2H), 4.23 (dd, J=11.0, 3.0 Hz, 1H), 3.93 SIRT-1 deacetylase fluorometric assay kit was purchased from (dd, J=11.0, 5.7 Hz, 1H), 3.39–3.32 (m, 1H), 2.92 (t, J=4.5 Hz, CycLex Ltd (Nagano, Japan).
Recommended publications
  • Phase I Trial of the PARP Inhibitor Olaparib and AKT Inhibitor Capivasertib in Patients with BRCA1/2- and Non–BRCA1/2-Mutant Cancers
    Published OnlineFirst June 12, 2020; DOI: 10.1158/2159-8290.CD-20-0163 RESEARCH ARTICLE Phase I Trial of the PARP Inhibitor Olaparib and AKT Inhibitor Capivasertib in Patients with BRCA1/2- and Non–BRCA1/2-Mutant Cancers Timothy A. Yap1,2, Rebecca Kristeleit3, Vasiliki Michalarea1, Stephen J. Pettitt4,5, Joline S.J. Lim1, Suzanne Carreira2, Desamparados Roda1,2, Rowan Miller3, Ruth Riisnaes2, Susana Miranda2, Ines Figueiredo2, Daniel Nava Rodrigues2, Sarah Ward1,2, Ruth Matthews1,2, Mona Parmar1,2, Alison Turner1,2, Nina Tunariu1, Neha Chopra1,4, Heidrun Gevensleben2, Nicholas C. Turner1,4, Ruth Ruddle2, Florence I. Raynaud2, Shaun Decordova2, Karen E. Swales2, Laura Finneran2, Emma Hall2, Paul Rugman6, Justin P.O. Lindemann6, Andrew Foxley6, Christopher J. Lord4,5, Udai Banerji1,2, Ruth Plummer7, Bristi Basu8, Juanita S. Lopez1,2, Yvette Drew7, and Johann S. de Bono1,2 Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst June 12, 2020; DOI: 10.1158/2159-8290.CD-20-0163 ABSTRACT Preclinical studies have demonstrated synergy between PARP and PI3K/AKT path- way inhibitors in BRCA1 and BRCA2 (BRCA1/2)–deficient andBRCA1/2 -proficient tumors. We conducted an investigator-initiated phase I trial utilizing a prospective intrapatient dose- escalation design to assess two schedules of capivasertib (AKT inhibitor) with olaparib (PARP inhibi- tor) in 64 patients with advanced solid tumors. Dose expansions enrolled germline BRCA1/2-mutant tumors, or BRCA1/2 wild-type cancers harboring somatic DNA damage response (DDR) or PI3K–AKT pathway alterations. The combination was well tolerated. Recommended phase II doses for the two schedules were: olaparib 300 mg twice a day with either capivasertib 400 mg twice a day 4 days on, 3 days off, or capivasertib 640 mg twice a day 2 days on, 5 days off.
    [Show full text]
  • The Influence of Cell Cycle Regulation on Chemotherapy
    International Journal of Molecular Sciences Review The Influence of Cell Cycle Regulation on Chemotherapy Ying Sun 1, Yang Liu 1, Xiaoli Ma 2 and Hao Hu 1,* 1 Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China; [email protected] (Y.S.); [email protected] (Y.L.) 2 Qingdao Institute of Measurement Technology, Qingdao 266000, China; [email protected] * Correspondence: [email protected] Abstract: Cell cycle regulation is orchestrated by a complex network of interactions between proteins, enzymes, cytokines, and cell cycle signaling pathways, and is vital for cell proliferation, growth, and repair. The occurrence, development, and metastasis of tumors are closely related to the cell cycle. Cell cycle regulation can be synergistic with chemotherapy in two aspects: inhibition or promotion. The sensitivity of tumor cells to chemotherapeutic drugs can be improved with the cooperation of cell cycle regulation strategies. This review presented the mechanism of the commonly used chemotherapeutic drugs and the effect of the cell cycle on tumorigenesis and development, and the interaction between chemotherapy and cell cycle regulation in cancer treatment was briefly introduced. The current collaborative strategies of chemotherapy and cell cycle regulation are discussed in detail. Finally, we outline the challenges and perspectives about the improvement of combination strategies for cancer therapy. Keywords: chemotherapy; cell cycle regulation; drug delivery systems; combination chemotherapy; cancer therapy Citation: Sun, Y.; Liu, Y.; Ma, X.; Hu, H. The Influence of Cell Cycle Regulation on Chemotherapy. Int. J. 1. Introduction Mol. Sci. 2021, 22, 6923. https:// Chemotherapy is currently one of the main methods of tumor treatment [1].
    [Show full text]
  • PARP Inhibitor Olaparib Causes No Potentiation of the Bleomycin Effect
    International Journal of Molecular Sciences Article PARP Inhibitor Olaparib Causes No Potentiation of the Bleomycin Effect in VERO Cells, Even in the Presence of Pooled ATM, DNA-PK, and LigIV Inhibitors Valentina Perini 1, Michelle Schacke 1, Pablo Liddle 1, Salomé Vilchez-Larrea 2 , Deborah J. Keszenman 3,* and Laura Lafon-Hughes 1,* 1 Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Departamento de Genética, Montevideo 11.600, Uruguay; [email protected] (V.P.); [email protected] (M.S.); [email protected] (P.L.) 2 Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires 1428, Argentina; [email protected] 3 Laboratorio de Radiobiología Médica y Ambiental, Grupo de Biofisicoquímica, Centro Universitario Regional Litoral Norte, Universidad de la República (UdelaR), Salto 50.000, Uruguay * Correspondence: [email protected] (D.J.K.); [email protected] (L.L.-H.) Received: 30 September 2020; Accepted: 19 October 2020; Published: 5 November 2020 Abstract: Poly(ADP-ribosyl)polymerase (PARP) synthesizes poly(ADP-ribose) (PAR), which is anchored to proteins. PAR facilitates multiprotein complexes’ assembly. Nuclear PAR affects chromatin’s structure and functions, including transcriptional regulation. In response to stress, particularly genotoxic stress, PARP activation facilitates DNA damage repair. The PARP inhibitor Olaparib (OLA) displays synthetic lethality with mutated homologous recombination proteins (BRCA-1/2), base excision repair proteins (XRCC1, Polβ), and canonical nonhomologous end joining (LigIV). However, the limits of synthetic lethality are not clear. On one hand, it is unknown whether any limiting factor of homologous recombination can be a synthetic PARP lethality partner.
    [Show full text]
  • Commissioner Final Decisions
    Commissioner for the Department for Medicaid Services Selections for Preferred Products This is a summary of the final Preferred Drug List (PDL) selections made by the Commissioner of the Department for Medicaid Services (DMS) based on the Drug Review and Options for Consideration document prepared for the Pharmacy and Therapeutics (P&T) Advisory Committee’s review on March 21, 2019, and the resulting official Committee recommendations. New Products to Market Epidiolex™ – Non-prefer in the PDL class: Anticonvulsants: Second Generation Length of Authorization: 1 year Epidiolex™ (cannabidiol), a non-psychoactive cannabinoid receptor antagonist, is approved for the treatment of seizures associated with Lennox-Gastaut syndrome or Dravet syndrome in patients ≥ 2 years of age. The mechanism by which cannabidiol exerts its anticonvulsant effects is unknown. Cannabidiol (Epidiolex) is a Schedule V controlled substance. Criteria for Approval: Diagnosis of Lennox-Gastaut syndrome (LGS) OR Dravet syndrome (DS); AND Prescriber is, or has a consultative relationship with, a neurology/epilepsy specialist; AND Trial and failure (e.g., incomplete seizure control) of at least 2 antiepileptic drugs; AND Must be used in adjunct with ≥ 1 antiepileptic drug. Age Limit: > 2 years Drug Class Preferred Agents Non-Preferred Agents Anticonvulsants: Banzel® CC, QL Briviact® QL Second Generation Gabitril® QL Epidiolex™ AE lamotrigine chewable tablets, tablets (except dose Fycompa™ QL packs) Keppra® tablets QL, solution levetiracetam solution, tablets QL Keppra XR® QL Sabril® CC Lamictal® topiramate QL Lamictal ODT® zonisamide QL Lamictal® XR™ QL lamotrigine dose packs lamotrigine ER QL lamotrigine ODT levetiracetam ER QL Qudexy® XR QL Spritam QL tiagabine QL Topamax® QL topiramate ER QL Trokendi XR™ QL vigabatrin Vimpat® QL AE – Age Edit; CC – Clinical Criteria; MD – Medications with Maximum Duration; QL – Quantity Limit; ST – Step Therapy © 2019, Magellan Medicaid Administration, a Magellan Rx Management company.
    [Show full text]
  • Candidate Synthetic Lethality Partners to PARP Inhibitors in the Treatment of Ovarian Clear Cell Cancer (Review)
    BIOMEDICAL REPORTS 7: 391-399, 2017 Candidate synthetic lethality partners to PARP inhibitors in the treatment of ovarian clear cell cancer (Review) NAOKI KAWAHARA, KENJI OGAWA, MIKA NAGAYASU, MAI KIMURA, YOSHIKAZU SASAKI and HIROSHI KOBAYASHI Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan Received August 18, 2017; Accepted September 14, 2017 DOI: 10.3892/br.2017.990 Abstract. Inhibitors of poly(ADP-ribose) polymerase Contents (PARP) are new types of personalized treatment of relapsed platinum-sensitive ovarian cancer harboring BRCA1/2 1. Introduction mutations. Ovarian clear cell cancer (CCC), a subset of 2. Systematic review of the literature using electronic ovarian cancer, often appears as low-stage disease with search in the PubMed/Medline databases a higher incidence among Japanese. Advanced CCC is 3. Future opportunities in the use of PARP inhibition in CCC highly aggressive with poor patient outcome. The aim of 4. Candidate mutated genes for enhancing the therapeutic the present study was to determine the potential synthetic ratio achieved by PARP inhibitors in CCC (Table IA). lethality gene pairs for PARP inhibitions in patients with 5. Upregulated genes enhancing synthetic lethality of CCC through virtual and biological screenings as well as PARP inhibitors in CCC (Table IB) clinical studies. We conducted a literature review for puta- 6. Synthetic lethal gene partners based on tive PARP sensitivity genes that are associated with the chemo resi stance-related genes in CCC (Table IC) CCC pathophysiology. Previous studies identified a variety 7. Discussion of putative target genes from several pathways associated with DNA damage repair, chromatin remodeling complex, PI3K-AKT-mTOR signaling, Notch signaling, cell cycle 1.
    [Show full text]
  • Negative Change Add Inclusion Criteria
    Policy Drug(s) Type of Change Brief Description of Policy Change new policy Elitek (rasburicase) n/a n/a new policy Retevmo (selpercatinib) n/a n/a new policy Tabrecta (capmatinib) n/a n/a new policy Trodelvy (govitecan-hziy) n/a n/a new policy Qinlock (ripretinib) n/a n/a UM ONC_1048 Campath (alemtuzumab) Archive Campath is no longer being used for any Hematology Oncology indications. Remove inclusion criteria: ALL: Induction, consolidation, maitenance, relapsed/refractory combination removed and use is supported as a part of a multi-agent chemotherapy regimen, for all sub-types of ALL, for induction/consolidation therapy, and for therapy of relapsed refractory disease NHL: Induction, consolidation, maitenance, relapsed/refractory combination removed and use is supported for extra- nodal NK/T-cell lymphoma (nasal type) and as a part of a multi-agent chemotherapy regimen for either first line therapy and/or therapy for relapsed /refractory disease. UM ONC_1063 Oncaspar (pegaspargase) Positive change Remove exclusion criteria: History of serious thrombosis, pancreatitis, or hemorrhagic events with L-asparaginase (ELSPAR) therapy. UM ONC_1064 Oncaspar (pegaspargase) Positive change Add exclusion criteria: Dosing exceeds single dose limit of Trastuzumab 8 mg/kg for the loading dose, 6 mg/kg for UM ONC_1134 Trastuzumab products Negative change subsequent doses when given every 3 weeks; 4 mg/kg for the loading dose and 2 mg/kg for the weekly dose. Remove exclusion criteria: Velcade (bortezomib) is being used after disease progression on another Velcade-based regimen. UM ONC_1136 Velcade (bortezomib) Positive change Add inclusion criteria: Multiple Myeloma: - NOTE: The preferred agent, per NCH policy, is generic bortezomib over brand bortezomib (Velcade) for all indications, unless there are contraindications or intolerance to generic bortezomib.
    [Show full text]
  • Rational Drug Combinations with PARP Inhibitors
    POTENTIAL CONFLICT OF INTEREST DISCLOSURES • Financial Relationships – SAB/Consultant: AstraZeneca, Catena Pharmaceuticals, Critical Outcome Technologies, ImmunoMET, Ionis, Medimmune, Nuevolution, Pfizer, Precision Medicine, Signalchem Lifesciences, Symphogen, Takeda/Millennium Pharmaceuticals, Tarveda, – Stock/ Options/Financial: Catena Pharmaceuticals, ImmunoMet, SignalChem, Spindle Top Ventures, Tarveda – Licensed Technology HRD assay to Myriad Genetics – Sponsored Research: Abbvie, Adelson Medical Research Foundation, AstraZeneca, Breast Cancer Research Foundation, Critical Outcomes Technology, Illumina, Ionis, Immunomet, Karus Therapeutics, Komen Research Foundation, Pfizer, Nanostring, Takeda/Millennium Pharmaceuticals, Tesaro I will discuss off label use and/or investigational use of drugs Dual mechanisms of action of PARPi Replication Fork Collapse PARP inhibitors Double strand breaks ADP ribosylation required for PARP to leave DNA Trapped PARP creates “toxic” double strand breaks Can PARP activity be extended beyond HRD PARP inhibitor responses are transient Ariel 2 Rucaparib Ian McNeish Lancet: LOH high is HRD assay performed by Foundation Med HRD BRCA mutant No detectable aberration Conclusion: Germline BRCA1/2 is strongest predictor of benefit HRD positivity identifies an additional population with significant benefit A population of patients without HRD show modest benefit Categorizing Predictive Biomarkers of Response for PARP inhibitors PARPness Deleterious gene variants or RNA/protein expression differences (e.g. SLFN11, E- Cadherin) not directly related to HRR deficiency that still engender PARP sensitivity. HRDness Increased genomic instability and reliance on error-prone DDR Loss of HRR efficiency Deleterious variants or post-translational loss of non-BRCA DDR genes (e.g. ATM), or select non-DDR genes (e.g. ARID1A); Hypoxia; Oncometabolites (e.g. 2-hydroxyglutarate). BRCAness Molecular phenocopy of tumors with BRCA1/2 deleterious mutations.
    [Show full text]
  • Low Levels of Circulating Estrogen Sensitize PTEN-Null Endometrial Tumors to PARP Inhibition in Vivo
    Published OnlineFirst November 12, 2013; DOI: 10.1158/1535-7163.MCT-13-0572 Molecular Cancer Cancer Therapeutics Insights Therapeutics Low Levels of Circulating Estrogen Sensitize PTEN-Null Endometrial Tumors to PARP Inhibition In Vivo Deanna M. Janzen1, Daniel Y. Paik1, Miguel A. Rosales1, Brian Yep1, Donghui Cheng8, Owen N. Witte2,4,7,8, Huseyin Kayadibi3, Christopher M. Ryan3, Michael E. Jung5, Kym Faull3, and Sanaz Memarzadeh1,6,7 Abstract Earlier in vitro work demonstrated that PARP inhibition induces cell death in PTEN-null endometrial cancer cell lines, but the in vivo therapeutic efficacy of these agents against endometrial cancer remains unknown. Here, we test the efficacy of AZD2281 (olaparib), an oral PARP inhibitor, in the therapy of PTEN-null endometrial tumors in a preclinical endometrial cancer mouse model. Primary endometrial tumors were generated by epithelial loss of PTEN using an in vivo model. This model recapitulates epithelial-specific loss of PTEN seen in human tumors, and histologically resembles endometrioid carcinomas, the predominant subtype of human endometrial cancers. Olaparib was administered orally to tumor-bearing mice in two hormonal extremes: high or low estrogen. Olaparib treatment achieved a significant reduction in tumor size in a low estrogenic milieu. In striking contrast, no response to olaparib was seen in tumors exposed to high levels of estrogen. Two key observations were made when estrogen levels were dropped: (i) the serum concentration of olaparib was significantly increased, resulting in sustained PARP inhibition at the tumor bed; and (ii) the homologous recombination pathway was compromised, as evidenced by decreased Rad51 protein expression and function. These two mechanisms may account for the sensitization of PTEN-null tumors to olaparib with estrogen deprivation.
    [Show full text]
  • Effects of Veliparib on Microglial Activation and Functional Outcomes After Traumatic Brain Injury in the Rat and Pig
    JOURNAL OF NEUROTRAUMA 35:918–929 (April 1, 2018) ª Mary Ann Liebert, Inc. DOI: 10.1089/neu.2017.5044 Effects of Veliparib on Microglial Activation and Functional Outcomes after Traumatic Brain Injury in the Rat and Pig Karen-Amanda Irvine,1 Robin K. Bishop,1 Seok Joon Won,1 Jianguo Xu,1,2 Katherine A. Hamel,3 Valerie Coppes,3 Pardeep Singh,1 Andrew Sondag,1 Eric Rome,1 Jayinee Basu,1 Giordano Fabricio Cittolin-Santos,1,4 S. Scott Panter,3 and Raymond A. Swanson1 Abstract The inflammation response induced by brain trauma can impair recovery. This response requires several hours to develop fully and thus provides a clinically relevant therapeutic window of opportunity. Poly(ADP-ribose) polymerase inhibitors suppress inflammatory responses, including brain microglial activation. We evaluated delayed treatment with veliparib, a poly(ADP- ribose) polymerase inhibitor, currently in clinical trials as a cancer therapeutic, in rats and pigs subjected to controlled cortical impact (CCI). In rats, CCI induced a robust inflammatory response at the lesion margins, scattered cell death in the dentate gyrus, and a delayed, progressive loss of corpus callosum axons. Pre-determined measures of cognitive and motor function showed evidence of attentional deficits that resolved after three weeks and motor deficits that recovered only partially over eight weeks. Veliparib was administered beginning 2 or 24 h after CCI and continued for up to 12 days. Veliparib suppressed CCI-induced microglial activation at doses of 3 mg/kg or higher and reduced reactive astrocytosis and cell death in the dentate gyrus, but had no significant effect on delayed axonal loss or functional recovery.
    [Show full text]
  • Lynparza Capsules Data Sheet
    1 NEW ZEALAND DATA SHEET 1. PRODUCT NAME LYNPARZA® 50 mg Capsules 2. QUALITATIVE AND QUANTITATIVE COMPOSITION Each capsule contains 50 mg of olaparib For the full list of excipients, see section 6.1. 3. PHARMACEUTICAL FORM Capsule White, opaque, size 0 hard capsule, marked with “OLAPARIB 50 mg” and the AstraZeneca logo in black ink. 4. CLINICAL PARTICULARS 4.1 THERAPEUTIC INDICATIONS Olaparib is indicated as monotherapy for the maintenance treatment of patients with platinum-sensitive relapsed BRCA-mutated (germline or somatic) high grade serous epithelial ovarian, fallopian tube or primary peritoneal cancer who are in response (complete response or partial response) after platinum-based chemotherapy. Prior treatment must have included at least 2 courses of platinum-based regimens. 4.2 DOSE AND METHOD OF ADMINISTRATION Treatment with LYNPARZA should be initiated and supervised by a physician experienced in the use of anticancer medicinal products. Important Administration Information LYNPARZA is also available as a 100mg and 150 mg tablet. DO NOT substitute LYNPARZA capsules (50 mg) with LYNPARZA tablets (100 mg and 150 mg) on a milligram-to-milligram basis due to differences in the dosing and bioavailability of each formulation. Refer to the Data Sheet for LYNPARZA tablets for specific tablet dosing. Patients must have confirmation of a breast cancer susceptibility gene (BRCA) mutation (germline or tumour) before LYNPARZA treatment is initiated. BRCA mutation status should be determined by an experienced laboratory using a validated test method. Dosage in adults The recommended dose of LYNPARZA is 400 mg (eight 50 mg capsules) taken twice daily, equivalent to a total daily dose of 800 mg.
    [Show full text]
  • FDA Listing of Established Pharmacologic Class Text Phrases January 2021
    FDA Listing of Established Pharmacologic Class Text Phrases January 2021 FDA EPC Text Phrase PLR regulations require that the following statement is included in the Highlights Indications and Usage heading if a drug is a member of an EPC [see 21 CFR 201.57(a)(6)]: “(Drug) is a (FDA EPC Text Phrase) indicated for Active Moiety Name [indication(s)].” For each listed active moiety, the associated FDA EPC text phrase is included in this document. For more information about how FDA determines the EPC Text Phrase, see the 2009 "Determining EPC for Use in the Highlights" guidance and 2013 "Determining EPC for Use in the Highlights" MAPP 7400.13.
    [Show full text]
  • Synergy Between the NAMPT Inhibitor GMX1777(8) and Pemetrexed in Non–Small Cell Lung Cancer Cells Is Mediated by PARP Activation and Enhanced NAD Consumption
    Published OnlineFirst August 21, 2014; DOI: 10.1158/0008-5472.CAN-14-0809 Cancer Priority Report Research Synergy between the NAMPT Inhibitor GMX1777(8) and Pemetrexed in Non–Small Cell Lung Cancer Cells Is Mediated by PARP Activation and Enhanced NAD Consumption Manuel Chan1, Michel Gravel1, Alexandre Bramoulle1,Gaelle€ Bridon2,3, Daina Avizonis2,3, Gordon C. Shore1, and Anne Roulston1 Abstract GMX1778 and its prodrug GMX1777 represent a new class of cancer drugs that targets nicotinamide phosphoribosyltransferase (NAMPT) as a new strategy to interfere with biosynthesis of the key enzymatic cofactor NAD, which is critical for a number of cell functions, including DNA repair. Using a genome-wide synthetic lethal siRNA screen, we identified the folate pathway–related genes, deoxyuridine triphosphatase and dihydrofolate reductase, the silencing of which sensitized non–small cell lung carcinoma (NSCLC) cells to the cytotoxic effects of GMX. Pemetrexed is an inhibitor of dihydrofolate reductase currently used to treat patients with nonsquamous NSCLC. We found that combining pemetrexed with GMX1777 produced a synergistic therapeutic benefit in A549 and H1299 NSCLC cells in vitro and in a mouse A549 xenograft model of lung cancer. Pemetrexed is known to activate PARPs, thereby accelerating NAD consumption. Genetic or pharma- cologic blockade of PARP activity inhibited this effect, impairing cell death by pemetrexed either alone or in combination with GMX1777. Conversely, inhibiting the base excision repair pathway accentuated NAD decline in response to GMX and the cytotoxicity of both agents either alone or in combination. These findings provide a mechanistic rationale for combining GMX1777 with pemetrexed as an effective new therapeutic strategy to treat nonsquamous NSCLC.
    [Show full text]